Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Brain ; 146(11): 4574-4593, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37678880

ABSTRACT

Neuromodulation by serotonin regulates the activity of neuronal networks responsible for a wide variety of essential behaviours. Serotonin (or 5-HT) typically activates metabotropic G protein-coupled receptors, which in turn initiate second messenger signalling cascades and induce short and long-lasting behavioural effects. Serotonin is intricately involved in the production of locomotor activity and gait control for different motor behaviours. Although dysfunction of serotonergic neurotransmission has been associated with mood disorders and spasticity after spinal cord injury, whether and to what extent such dysregulation is implicated in movement disorders has not been firmly established. Here, we investigated whether serotonergic neuromodulation is affected in spinal muscular atrophy (SMA), a neurodegenerative disease caused by ubiquitous deficiency of the SMN protein. The hallmarks of SMA are death of spinal motor neurons, muscle atrophy and impaired motor control, both in human patients and mouse models of disease. We used a severe mouse model of SMA, that closely recapitulates the severe symptoms exhibited by type I SMA patients, the most common and most severe form of the disease. Together, with mouse genetics, optogenetics, physiology, morphology and behavioural analysis, we report severe dysfunction of serotonergic neurotransmission in the spinal cord of SMA mice, both at early and late stages of the disease. This dysfunction is followed by reduction of 5-HT synapses on vulnerable motor neurons. We demonstrate that motor neurons innervating axial and trunk musculature are preferentially affected, suggesting a possible cause for the proximo-distal progression of disease, and raising the possibility that it may underlie scoliosis in SMA patients. We also demonstrate that the 5-HT dysfunction is caused by SMN deficiency in serotonergic neurons in the raphe nuclei of the brainstem. The behavioural significance of the dysfunction in serotonergic neuromodulation is underlined by inter-limb discoordination in SMA mice, which is ameliorated when selective restoration of SMN in 5-HT neurons is achieved by genetic means. Our study uncovers an unexpected dysfunction of serotonergic neuromodulation in SMA and indicates that, if normal function is to be restored under disease conditions, 5-HT neuromodulation should be a key target for therapeutic approaches.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Humans , Mice , Animals , Serotonin/metabolism , Neurodegenerative Diseases/metabolism , Muscular Atrophy, Spinal/genetics , Motor Neurons/metabolism , Spinal Cord/metabolism , Disease Models, Animal
2.
Adv Neurobiol ; 28: 63-85, 2022.
Article in English | MEDLINE | ID: mdl-36066821

ABSTRACT

This chapter will provide an introduction into motoneuron anatomy, electrophysiological properties, firing patterns focusing on development and also describing several pathological conditions that affect mononeurons. It starts with a historical retrospective describing the early landmark work into motoneurons. The next section lays out the various types of motoneurons (alpha, beta, and gamma) and their subclasses (fast-twitch fatigable, fast-twitch fatigue-resistant, and slow-twitch fatigue resistant), highlighting the functional relevance of this classification scheme. The third section describes the development of motoneurons' passive and active electrophysiological properties. This section also defines the major terms one uses in describing how a neuron functions electrophysiologically. The electrophysiological aspects of a neuron is critical to understanding how it behaves within a circuit and contributes to behavior since the firing of an action potential is how neurons communicate with each other and with muscles. The electrophysiological changes of motoneurons over development underlies how their function changes over the lifetime of an organism. After describing the properties of individual motoneurons, the chapter then turns to revealing how motoneurons interact within complex neural circuits, with other motoneurons as well as sensory neurons, and how these circuits change over development. Finally, this chapter ends with highlighting some recent advances made in motoneuron pathology, focusing on spinal muscular atrophy, amyotrophic lateral sclerosis, and axotomy.


Subject(s)
Amyotrophic Lateral Sclerosis , Muscular Atrophy, Spinal , Amyotrophic Lateral Sclerosis/pathology , Humans , Motor Neurons , Muscles , Retrospective Studies
3.
Cell Rep ; 40(12): 111393, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130491

ABSTRACT

The neuromuscular junction (NMJ) is an essential synapse whose loss is a key hallmark of the neurodegenerative disease spinal muscular atrophy (SMA). Here, we show that activity of the SMA-determining SMN protein in the assembly of U7 small nuclear ribonucleoprotein (snRNP)-which functions in the 3'-end processing of replication-dependent histone mRNAs-is required for NMJ integrity. Co-expression of U7-specific Lsm10 and Lsm11 proteins selectively enhances U7 snRNP assembly, corrects histone mRNA processing defects, and rescues key structural and functional abnormalities of neuromuscular pathology in SMA mice-including NMJ denervation, decreased synaptic transmission, and skeletal muscle atrophy. Furthermore, U7 snRNP dysfunction drives selective loss of the synaptic organizing protein Agrin at NMJs innervating vulnerable muscles of SMA mice. These findings reveal a direct contribution of U7 snRNP dysfunction to neuromuscular pathology in SMA and suggest a role for histone gene regulation in maintaining functional synaptic connections between motor neurons and muscles.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Agrin/metabolism , Animals , Histones/metabolism , Mice , Muscular Atrophy, Spinal/metabolism , Neurodegenerative Diseases/metabolism , Neuromuscular Junction/metabolism , RNA, Messenger/metabolism , Ribonucleoprotein, U7 Small Nuclear/chemistry , Ribonucleoprotein, U7 Small Nuclear/metabolism
4.
STAR Protoc ; 3(1): 101236, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35300003

ABSTRACT

Loss of synapses on spinal motor neurons is a major feature of several neurodegenerative diseases; however, analyzing these premotor synapses is challenging because of their small size and high density. This protocol describes confocal and Stimulated Emission Depletion (STED) imaging of murine spinal premotor synapses and their segment-specific quantification by confocal microscopy. We detail the preparation of spinal cord segments, followed by image acquisition and analysis. This protocol enables in-depth analysis of pathological changes in spinal premotor synapses during neurodegeneration. For complete details on the use and execution of this protocol, please refer to Buettner et al. (2021).


Subject(s)
Neurodegenerative Diseases , Spinal Cord , Animals , Mice , Microscopy, Confocal , Motor Neurons , Spinal Cord/diagnostic imaging , Synapses
5.
Cell ; 185(2): 328-344.e26, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35063074

ABSTRACT

Locomotion is a complex behavior required for animal survival. Vertebrate locomotion depends on spinal interneurons termed the central pattern generator (CPG), which generates activity responsible for the alternation of flexor and extensor muscles and the left and right side of the body. It is unknown whether multiple or a single neuronal type is responsible for the control of mammalian locomotion. Here, we show that ventral spinocerebellar tract neurons (VSCTs) drive generation and maintenance of locomotor behavior in neonatal and adult mice. Using mouse genetics, physiological, anatomical, and behavioral assays, we demonstrate that VSCTs exhibit rhythmogenic properties and neuronal circuit connectivity consistent with their essential role in the locomotor CPG. Importantly, optogenetic activation and chemogenetic silencing reveals that VSCTs are necessary and sufficient for locomotion. These findings identify VSCTs as critical components for mammalian locomotion and provide a paradigm shift in our understanding of neural control of complex behaviors.


Subject(s)
Locomotion/physiology , Mammals/physiology , Motor Neurons/cytology , Spinocerebellar Tracts/cytology , Animals , Axons/physiology , Electrophysiological Phenomena , Gap Junctions/metabolism , Gene Silencing , Glutamic Acid/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Interneurons/physiology , Lumbar Vertebrae/metabolism , Mice , Proprioception , Swimming , Synapses/physiology , Transcription Factors/metabolism
6.
Nat Commun ; 12(1): 5040, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413305

ABSTRACT

SMN is a ubiquitously expressed protein and is essential for life. SMN deficiency causes the neurodegenerative disease spinal muscular atrophy (SMA), the leading genetic cause of infant mortality. SMN interacts with itself and other proteins to form a complex that functions in the assembly of ribonucleoproteins. SMN is modified by SUMO (Small Ubiquitin-like Modifier), but whether sumoylation is required for the functions of SMN that are relevant to SMA pathogenesis is not known. Here, we show that inactivation of a SUMO-interacting motif (SIM) alters SMN sub-cellular distribution, the integrity of its complex, and its function in small nuclear ribonucleoproteins biogenesis. Expression of a SIM-inactivated mutant of SMN in a mouse model of SMA slightly extends survival rate with limited and transient correction of motor deficits. Remarkably, although SIM-inactivated SMN attenuates motor neuron loss and improves neuromuscular junction synapses, it fails to prevent the loss of sensory-motor synapses. These findings suggest that sumoylation is important for proper assembly and function of the SMN complex and that loss of this post-translational modification impairs the ability of SMN to correct selective deficits in the sensory-motor circuit of SMA mice.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Neurodegenerative Diseases/pathology , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism , Sumoylation , Synapses/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Disease Models, Animal , Humans , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Neurodegenerative Diseases/metabolism , Synapses/pathology , Zebrafish
7.
Nat Neurosci ; 24(7): 930-940, 2021 07.
Article in English | MEDLINE | ID: mdl-33795885

ABSTRACT

The neurodegenerative disease spinal muscular atrophy (SMA) is caused by deficiency in the survival motor neuron (SMN) protein. Currently approved SMA treatments aim to restore SMN, but the potential for SMN expression beyond physiological levels is a unique feature of adeno-associated virus serotype 9 (AAV9)-SMN gene therapy. Here, we show that long-term AAV9-mediated SMN overexpression in mouse models induces dose-dependent, late-onset motor dysfunction associated with loss of proprioceptive synapses and neurodegeneration. Mechanistically, aggregation of overexpressed SMN in the cytoplasm of motor circuit neurons sequesters components of small nuclear ribonucleoproteins, leading to splicing dysregulation and widespread transcriptome abnormalities with prominent signatures of neuroinflammation and the innate immune response. Thus, long-term SMN overexpression interferes with RNA regulation and triggers SMA-like pathogenic events through toxic gain-of-function mechanisms. These unanticipated, SMN-dependent and neuron-specific liabilities warrant caution on the long-term safety of treating individuals with SMA with AAV9-SMN and the risks of uncontrolled protein expression by gene therapy.


Subject(s)
Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration , Survival of Motor Neuron 1 Protein/toxicity , Animals , Dependovirus , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Injections, Intraventricular , Mice , Motor Disorders/genetics , Motor Disorders/metabolism , Motor Disorders/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Survival of Motor Neuron 1 Protein/genetics
9.
Nat Commun ; 12(1): 1451, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649316

ABSTRACT

Proprioceptive feedback mainly derives from groups Ia and II muscle spindle (MS) afferents and group Ib Golgi tendon organ (GTO) afferents, but the molecular correlates of these three afferent subtypes remain unknown. We performed single cell RNA sequencing of genetically identified adult proprioceptors and uncovered five molecularly distinct neuronal clusters. Validation of cluster-specific transcripts in dorsal root ganglia and skeletal muscle demonstrates that two of these clusters correspond to group Ia MS afferents and group Ib GTO afferent proprioceptors, respectively, and suggest that the remaining clusters could represent group II MS afferents. Lineage analysis between proprioceptor transcriptomes at different developmental stages provides evidence that proprioceptor subtype identities emerge late in development. Together, our data provide comprehensive molecular signatures for groups Ia and II MS afferents and group Ib GTO afferents, enabling genetic interrogation of the role of individual proprioceptor subtypes in regulating motor output.


Subject(s)
Mechanoreceptors/metabolism , Muscle Spindles/metabolism , Neurons, Afferent/metabolism , Animals , Calbindin 2/metabolism , Electrophysiological Phenomena , Ion Channels/metabolism , Mice, Transgenic , Neurons/metabolism , Proprioception , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Neurotransmitter/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics
10.
Sci Transl Med ; 13(578)2021 01 27.
Article in English | MEDLINE | ID: mdl-33504650

ABSTRACT

Gene replacement and pre-mRNA splicing modifier therapies represent breakthrough gene targeting treatments for the neuromuscular disease spinal muscular atrophy (SMA), but mechanisms underlying variable efficacy of treatment are incompletely understood. Our examination of severe infantile onset human SMA tissues obtained at expedited autopsy revealed persistence of developmentally immature motor neuron axons, many of which are actively degenerating. We identified similar features in a mouse model of severe SMA, in which impaired radial growth and Schwann cell ensheathment of motor axons began during embryogenesis and resulted in reduced acquisition of myelinated axons that impeded motor axon function neonatally. Axons that failed to ensheath degenerated rapidly postnatally, specifically releasing neurofilament light chain protein into the blood. Genetic restoration of survival motor neuron protein (SMN) expression in mouse motor neurons, but not in Schwann cells or muscle, improved SMA motor axon development and maintenance. Treatment with small-molecule SMN2 splice modifiers beginning immediately after birth in mice increased radial growth of the already myelinated axons, but in utero treatment was required to restore axonal growth and associated maturation, prevent subsequent neonatal axon degeneration, and enhance motor axon function. Together, these data reveal a cellular basis for the fulminant neonatal worsening of patients with infantile onset SMA and identify a temporal window for more effective treatment. These findings suggest that minimizing treatment delay is critical to achieve optimal therapeutic efficacy.


Subject(s)
Muscular Atrophy, Spinal , Animals , Axons , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Neurons , Muscular Atrophy, Spinal/therapy , Survival of Motor Neuron 1 Protein/genetics
11.
J Neurosci ; 41(2): 376-389, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33219005

ABSTRACT

Dysfunction of neuronal circuits is an important determinant of neurodegenerative diseases. Synaptic dysfunction, death, and intrinsic activity of neurons are thought to contribute to the demise of normal behavior in the disease state. However, the interplay between these major pathogenic events during disease progression is poorly understood. Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by a deficiency in the ubiquitously expressed protein SMN and is characterized by motor neuron death, skeletal muscle atrophy, as well as dysfunction and loss of both central and peripheral excitatory synapses. These disease hallmarks result in an overall reduction of neuronal activity in the spinal sensory-motor circuit. Here, we show that increasing neuronal activity by chronic treatment with the FDA-approved potassium channel blocker 4-aminopyridine (4-AP) improves motor behavior in both sexes of a severe mouse model of SMA. 4-AP restores neurotransmission and number of proprioceptive synapses and neuromuscular junctions (NMJs), while having no effects on motor neuron death. In addition, 4-AP treatment with pharmacological inhibition of p53-dependent motor neuron death results in additive effects, leading to full correction of sensory-motor circuit pathology and enhanced phenotypic benefit in SMA mice. Our in vivo study reveals that 4-AP-induced increase of neuronal activity restores synaptic connectivity and function in the sensory-motor circuit to improve the SMA motor phenotype.SIGNIFICANCE STATEMENT Spinal muscular atrophy (SMA) is a neurodegenerative disease, characterized by synaptic loss, motor neuron death, and reduced neuronal activity in spinal sensory-motor circuits. However, whether these are parallel or dependent events is unclear. We show here that long-term increase of neuronal activity by the FDA-approved drug 4-aminopyridine (4-AP) rescues the number and function of central and peripheral synapses in a SMA mouse model, resulting in an improvement of the sensory-motor circuit and motor behavior. Combinatorial treatment of pharmacological inhibition of p53, which is responsible for motor neuron death and 4-AP, results in additive beneficial effects on the sensory-motor circuit in SMA. Thus, neuronal activity restores synaptic connections and improves significantly the severe SMA phenotype.


Subject(s)
Movement Disorders/drug therapy , Muscular Atrophy, Spinal/drug therapy , Psychomotor Performance/drug effects , Sensation Disorders/drug therapy , 4-Aminopyridine/therapeutic use , Animals , Cell Death/drug effects , Mice , Mice, Knockout , Motor Neurons/drug effects , Movement Disorders/etiology , Movement Disorders/psychology , Muscular Atrophy, Spinal/complications , Muscular Atrophy, Spinal/psychology , Neuromuscular Junction/drug effects , Potassium Channel Blockers/therapeutic use , Proprioception/drug effects , Sensation Disorders/etiology , Sensation Disorders/psychology , Survival of Motor Neuron 1 Protein/genetics , Synapses/drug effects , Synaptic Transmission/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors
13.
Cell Rep ; 29(12): 3885-3901.e5, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851921

ABSTRACT

Reduced expression of the survival motor neuron (SMN) protein causes the neurodegenerative disease spinal muscular atrophy (SMA). Here, we show that adeno-associated virus serotype 9 (AAV9)-mediated delivery of Stasimon-a gene encoding an endoplasmic reticulum (ER)-resident transmembrane protein regulated by SMN-improves motor function in a mouse model of SMA through multiple mechanisms. In proprioceptive neurons, Stasimon overexpression prevents the loss of afferent synapses on motor neurons and enhances sensory-motor neurotransmission. In motor neurons, Stasimon suppresses neurodegeneration by reducing phosphorylation of the tumor suppressor p53. Moreover, Stasimon deficiency converges on SMA-related mechanisms of p53 upregulation to induce phosphorylation of p53 through activation of p38 mitogen-activated protein kinase (MAPK), and pharmacological inhibition of this kinase prevents motor neuron death in SMA mice. These findings identify Stasimon dysfunction induced by SMN deficiency as an upstream driver of distinct cellular cascades that lead to synaptic loss and motor neuron degeneration, revealing a dual contribution of Stasimon to motor circuit pathology in SMA.


Subject(s)
Membrane Proteins/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/etiology , Sensory Receptor Cells/pathology , Survival of Motor Neuron 1 Protein/physiology , Synapses/pathology , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Dependovirus/genetics , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Mice , Mice, Knockout , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Sensory Receptor Cells/metabolism , Synapses/metabolism , Tumor Suppressor Protein p53/genetics , p38 Mitogen-Activated Protein Kinases/genetics
14.
Cell Rep ; 29(10): 3087-3100.e7, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801075

ABSTRACT

Movement is an essential behavior requiring the assembly and refinement of spinal motor circuits. However, the mechanisms responsible for circuit refinement and synapse maintenance are poorly understood. Similarly, the molecular mechanisms by which gene mutations cause dysfunction and elimination of synapses in neurodegenerative diseases that occur during development are unknown. Here, we demonstrate that the complement protein C1q is required for the refinement of sensory-motor circuits during normal development, as well as for synaptic dysfunction and elimination in spinal muscular atrophy (SMA). C1q tags vulnerable SMA synapses, which triggers activation of the classical complement pathway leading to microglia-mediated elimination. Pharmacological inhibition of C1q or depletion of microglia rescues the number and function of synapses, conferring significant behavioral benefit in SMA mice. Thus, the classical complement pathway plays critical roles in the refinement of developing motor circuits, while its aberrant activation contributes to motor neuron disease.


Subject(s)
Complement Pathway, Classical/physiology , Microglia/metabolism , Muscular Atrophy, Spinal/metabolism , Animals , Child, Preschool , Complement C1q/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Synapses/metabolism
15.
Neuron ; 100(1): 135-149.e7, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30308166

ABSTRACT

Neuronal diversification is a fundamental step in the construction of functional neural circuits, but how neurons generated from single progenitor domains acquire diverse subtype identities remains poorly understood. Here we developed an embryonic stem cell (ESC)-based system to model subtype diversification of V1 interneurons, a class of spinal neurons comprising four clades collectively containing dozens of molecularly distinct neuronal subtypes. We demonstrate that V1 subtype diversity can be modified by extrinsic signals. Inhibition of Notch and activation of retinoid signaling results in a switch to MafA clade identity and enriches differentiation of Renshaw cells, a specialized MafA subtype that mediates recurrent inhibition of spinal motor neurons. We show that Renshaw cells are intrinsically programmed to migrate to species-specific laminae upon transplantation and to form subtype-specific synapses with motor neurons. Our results demonstrate that stem cell-derived neuronal subtypes can be used to investigate mechanisms underlying neuronal subtype specification and circuit assembly.


Subject(s)
Interneurons/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Synapses/metabolism , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Interneurons/metabolism , Mice , Motor Neurons/cytology , Motor Neurons/metabolism , Neural Stem Cells/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism
16.
Genes Dev ; 32(15-16): 1045-1059, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30012555

ABSTRACT

Ubiquitous deficiency in the survival motor neuron (SMN) protein causes death of motor neurons-a hallmark of the neurodegenerative disease spinal muscular atrophy (SMA)-through poorly understood mechanisms. Here, we show that the function of SMN in the assembly of spliceosomal small nuclear ribonucleoproteins (snRNPs) regulates alternative splicing of Mdm2 and Mdm4, two nonredundant repressors of p53. Decreased inclusion of critical Mdm2 and Mdm4 exons is most prominent in SMA motor neurons and correlates with both snRNP reduction and p53 activation in vivo. Importantly, increased skipping of Mdm2 and Mdm4 exons regulated by SMN is necessary and sufficient to synergistically elicit robust p53 activation in wild-type mice. Conversely, restoration of full-length Mdm2 and Mdm4 suppresses p53 induction and motor neuron degeneration in SMA mice. These findings reveal that loss of SMN-dependent regulation of Mdm2 and Mdm4 alternative splicing underlies p53-mediated death of motor neurons in SMA, establishing a causal link between snRNP dysfunction and neurodegeneration.


Subject(s)
Alternative Splicing , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Death , Exons , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/physiopathology , NIH 3T3 Cells , Nerve Degeneration/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Ribonucleoproteins, Small Nuclear/biosynthesis , Tumor Suppressor Protein p53/metabolism
17.
Elife ; 72018 02 20.
Article in English | MEDLINE | ID: mdl-29460776

ABSTRACT

In amyotrophic lateral sclerosis (ALS) and animal models of ALS, including SOD1-G93A mice, disassembly of the neuromuscular synapse precedes motor neuron loss and is sufficient to cause a decline in motor function that culminates in lethal respiratory paralysis. We treated SOD1-G93A mice with an agonist antibody to MuSK, a receptor tyrosine kinase essential for maintaining neuromuscular synapses, to determine whether increasing muscle retrograde signaling would slow nerve terminal detachment from muscle. The agonist antibody, delivered after disease onset, slowed muscle denervation, promoting motor neuron survival, improving motor system output, and extending the lifespan of SOD1-G93A mice. These findings suggest a novel therapeutic strategy for ALS, using an antibody format with clinical precedence, which targets a pathway essential for maintaining attachment of nerve terminals to muscle.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Antibodies/administration & dosage , Immunologic Factors/administration & dosage , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Disease Models, Animal , Mice , Motor Neurons/drug effects , Motor Neurons/physiology , Treatment Outcome
18.
Cell Rep ; 21(13): 3767-3780, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29281826

ABSTRACT

The hallmark of spinal muscular atrophy (SMA), an inherited disease caused by ubiquitous deficiency in the SMN protein, is the selective degeneration of subsets of spinal motor neurons. Here, we show that cell-autonomous activation of p53 occurs in vulnerable but not resistant motor neurons of SMA mice at pre-symptomatic stages. Moreover, pharmacological or genetic inhibition of p53 prevents motor neuron death, demonstrating that induction of p53 signaling drives neurodegeneration. At late disease stages, however, nuclear accumulation of p53 extends to resistant motor neurons and spinal interneurons but is not associated with cell death. Importantly, we identify phosphorylation of serine 18 as a specific post-translational modification of p53 that exclusively marks vulnerable SMA motor neurons and provide evidence that amino-terminal phosphorylation of p53 is required for the neurodegenerative process. Our findings indicate that distinct events induced by SMN deficiency converge on p53 to trigger selective death of vulnerable SMA motor neurons.


Subject(s)
Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers/metabolism , Cell Death , Female , Male , Mice , Models, Biological , Phosphorylation
19.
Nat Neurosci ; 20(7): 905-916, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28504671

ABSTRACT

Behavioral deficits in neurodegenerative diseases are often attributed to the selective dysfunction of vulnerable neurons via cell-autonomous mechanisms. Although vulnerable neurons are embedded in neuronal circuits, the contributions of their synaptic partners to disease process are largely unknown. Here we show that, in a mouse model of spinal muscular atrophy (SMA), a reduction in proprioceptive synaptic drive leads to motor neuron dysfunction and motor behavior impairments. In SMA mice or after the blockade of proprioceptive synaptic transmission, we observed a decrease in the motor neuron firing that could be explained by the reduction in the expression of the potassium channel Kv2.1 at the surface of motor neurons. Chronically increasing neuronal activity pharmacologically in vivo led to a normalization of Kv2.1 expression and an improvement in motor function. Our results demonstrate a key role of excitatory synaptic drive in shaping the function of motor neurons during development and the contribution of its disruption to a neurodegenerative disease.


Subject(s)
Motor Neurons/physiology , Muscular Atrophy, Spinal/physiopathology , Proprioception/physiology , Shab Potassium Channels/physiology , Synapses/physiology , Action Potentials/physiology , Animals , Cell Survival/physiology , Disease Models, Animal , Kainic Acid/pharmacology , Metalloendopeptidases/pharmacology , Mice , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/metabolism , Neuromuscular Junction/physiology , Reflex, Righting/physiology , Shab Potassium Channels/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Synapses/drug effects , Tetanus Toxin/pharmacology
20.
Cell Rep ; 16(5): 1416-1430, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27452470

ABSTRACT

In spinal muscular atrophy, a neurodegenerative disease caused by ubiquitous deficiency in the survival motor neuron (SMN) protein, sensory-motor synaptic dysfunction and increased excitability precede motor neuron (MN) loss. Whether central synaptic dysfunction and MN hyperexcitability are cell-autonomous events or they contribute to MN death is unknown. We addressed these issues using a stem-cell-based model of the motor circuit consisting of MNs and both excitatory and inhibitory interneurons (INs) in which SMN protein levels are selectively depleted. We show that SMN deficiency induces selective MN death through cell-autonomous mechanisms, while hyperexcitability is a non-cell-autonomous response of MNs to defects in pre-motor INs, leading to loss of glutamatergic synapses and reduced excitation. Findings from our in vitro model suggest that dysfunction and loss of MNs result from differential effects of SMN deficiency in distinct neurons of the motor circuit and that hyperexcitability does not trigger MN death.


Subject(s)
Cell Survival/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Stem Cells/metabolism , Stem Cells/pathology , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Line , Disease Models, Animal , Interneurons/metabolism , Interneurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Synapses/metabolism , Synapses/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...